Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jun 15.
Published in final edited form as: Annu Rev Microbiol. 2018 Jun 21;72:399–422. doi: 10.1146/annurev-micro-090817-062307

Communication between the microbiota and mammalian immunity

Kyla Ost 1, June L Round 1,*
PMCID: PMC7294967  NIHMSID: NIHMS1586544  PMID: 29927706

Abstract

Mammalian immune systems evolved within a diverse and dominant microbial world, forcing reciprocal interactions between the two. Adaptive immunity enacts specific responses against microbes. These antigen specific responses have classically been studied in the context of pathogenicity, however are now known to have significant effects on our resident microbes. In turn, microbes employ an arsenal of mechanisms to influence development and specificity of host immunity. Understanding these complex reactions will be necessary to develop microbiota based therapies to prevent or treat disease. Here we review the literature detailing the cross-talk between resident microbes with a focus on the specificity of the host response and the microbial molecules that influence them.

Specificity of immune responses toward commensals

The GI tract harbors a highly complex microbial community that actively communicates with the intestinal immune system, significantly impacting host health. The adaptive arm of the immune system, comprised of T and B lymphocytes, interact with this microbial community to both prevent microbial invasion and pathogenesis while also preventing detrimental immune responses toward commensal microbes. Classically, T and B cells react to foreign molecules, or antigens, via their T and B cell receptors, and therefore their responses are considered to be highly specific to target precise organisms. Within the intestine there is a vast amount of dietary and microbial antigens that can continuously stimulate intestinal lymphocytes. This constant stimulation promotes immune processes to maintain a beneficial interaction with the microbiota, but can also promote pathogenic inflammatory responses. Whether these commensal induced responses are specific to the gut bacteria themselves or result in “bystander” immunity is still being evaluated. A number of reviews have recently and comprehensively described the current literature that summarizes the generalized immune responses that are induced by commensal microbes (1, 2). In this section, we will analyze the cell types and specificity of these responses.

Dictating specificity: Major histocompatibility complex (MHC)

T cells detect antigens presented by glycoproteins encoded by major histocompatibility genes (MHC). T cells express co-receptors, CD8 and CD4, that interact with MHC I and II, respectively. MHC presentation of antigens to T cells serves as the basis for initiating antigen specificity. Several studies have demonstrated that the composition of the microbiota is dependent on the types of MHC alleles expressed by the host (37). MHC genes are some of the most diverse loci in the human genome and different alleles have distinct antigen presentation properties. As MHC/T cell interactions are the basis for all adaptive immunity, it is not surprising that MHC genetic variation impacts susceptibility to infectious diseases and autoimmune diseases (8). One of the mechanisms for this was recently attributed to how MHC influences assembly of the microbiota. Using mice expressing distinct MHC loci, we demonstrated that resident microbial communities were shaped differently depending on the MHC expressed. Different MHCll alleles altered antigen-specific antibody (IgA) responses in the gut, resulting in distinct microbial communities. These communities provided significantly different levels of protection against the enteric pathogen, Salmonella typhimurium (4).

Recently, MHC/microbiota interaction has been shown to protect mice against type I diabetes (T1D). In humans, MHC variation is the strongest genetic variant linked to T1D (8). Consistent with humans, the expression of a particular MHC allele (Eα complex) in the non-obese diabetic (NOD) mouse model of T1D protected mice from disease (9). The mechanism had been a mystery until recently when Silverman and colleges showed that this protection is mediated by the intestinal microbiota. Their first clue was that NOD mothers expressing the protective allele could transfer protection to their offspring without the allele. They then demonstrated that different antibiotics ameliorated the protective effect (6), demonstrating that commensal bacteria underlie this protection. These recent studies offer the first mechanistic evidence that diseases affected by MHC genetic variation may be regulated by microbiota alterations.

Antigen acquisition and antigen presenting cells.

A network of antigen presenting cells are responsible for acquiring and coordinating antigen-specific immune responses and mononuclear phagocytes, particularly dendritic cells, are central to this process (Figure 1). Dendritic cells are found throughout the intestine, but are concentrated in the lamina propria and lymphoid tissues that include Peyer’s patches and isolated lymphoid follicles in the small intestine and the draining mesenteric lymph node. There are 3 main DC groups found in the gut and they are differentiated by the expression of three surface markers, the αECD103 integrin, CD11b and CX3CR1 (10). In the small intestine, the majority of the DCs are CD103+CD11b+ and are located in both the lamina propria and CD103+CD11b− are primarily located in Peyer’s Patches (11). These dendritic cells are migratory, constantly sampling both dietary and commensal antigens and trafficking them to the MLN to activate T cells (12, 13).

Figure 1: Important antigen-presenting cells in the gut.

Figure 1:

The main DC populations in the gut are CD103+CD11b−, CD103+CD11b+, and CD103−CD11b+CX3CR1+ DCs. CD103+ DCs acquire antigens through specialized M cells, transport through goblet cells, invading microbes, and even from CD103−CD11b+CX3CR1+ DCs. These DCs constitutively express CCR7 and transport antigens to the MLN for T cell activation. CD103−CD11b+CX3CR1+ DCs can directly sample intestinal microbes by sending cellular projections through the epithelial cell layer. CD103−CD11b+CX3CR1+ DCs are not migratory under normal conditions, but upregulate CCR7 and migrate to the MLN after antibiotic-disruption of the microbiota, or in MyD88−/− mutant mice. These DC populations are required for inducing different T cell responses. CD103+ DCs are particularly good at inducing Treg differentiation. CD103+CD11b− DCs are required for Th1 responses, while CD103+CD11b− DCs are required for Th17 responses. CD103−CD11b+CX3CR1+ DCs are able to induce both Th1 and Th17 responses, and are required for SFB-specific Th17 responses. Finally, CCR6 expressing ILC3 cells suppress commensal-specific T cells by presenting MHCII-bound commensal antigens and inducing T cell apoptosis.

There are a number of pathways that deliver antigens to these DCs. Mucus secreting goblet cells transports low molecular weight antigens to CD103+ DCs in the small intestine (14), specialized M cells overlaying Peyer’s patches allow passage of antigens and microbes to Peyer’s patch DCs, and pathogens that breach the epithelial cell layer. CD103+ CD11b+ DCs express high levels of the CCR7, which is required for migration of these DCs from mucosal tissues to the MLN (12, 15). CCR7 expressing DCs have also been shown to transport commensal microbes from the intestine to the MLN to stimulate commensal antigen-specific responses (16).

In the colon, the primary DC population is CD103−CD11b+ and also expresses the fractalkin receptor CX3CR1, though they also have important functions in the SI (17). Unlike CD103+ DCs, these cells are macrophage-like and arise from monocyte progenitors (12, 18) and been shown acquire antigen in ways distinct from CD103+ cells. Niess et al. used GFP-CX3CR5 mouse models to show that these cells extend cellular projections from the lamina propria, between epithelial cells, and into the intestinal lumen to sample microbial antigens, a process that required CX3CR1 expression (19). Unlike CD103+ DCs, CD103−CD11b+CX3CR1+ require microbial signals for development and are depleted in germ free mice (20). Also distinct from CD103+ DCs, these macrophage-like DCs do not express high levels of CCR7 and do not traffic to the MLN under homeostatic conditions (12). However, Diehl et al. demonstrated that these cells upregulated CCR7 and migrate to the MLN when the microbiota is depleted by antibiotics. This suggests microbial signals suppress CCR7 expression on these DCs, preventing migration, and potentially aberrant immune activation (21). CD103+ and CD103−CD11b+CX3CR1+ DC populations have also been shown to cooperate to acquire them to the MLN and CD103−CD11b+ CX3CR1+ DCs were shown to transfer antigens to CD103+ DCs, which transport them to the MLN (22).

Interestingly, there are inherent differences in the way these DC groups activate T cells (Figure 1). For example, CD103+ DCs are particularly adept at inducing Treg differentiation by secreting Treg inducing molecules, including retinoic acid and TGF-β (23). Specific DCs also maintain different T helper cell populations in the gut. CD103+CD11b+ DCs are required for maintaining Th17 cells, while CD103+CD11b− DCs were recently demonstrated to maintain Th1 responses (2426). CD103−CD11b+ CX3CR1 DCs induce both Th1 and Th17 responses (21, 24).

A non-classical antigen presenting cell that is important for mediating T cell responses are innate lymphoid cells (ILCs) (27). Particular subset of ILCs, called CCR6 ILC3, express MHCII that serve an important regulatory role in intestinal immune homeostasis. Selective deletion of MHCII in CCR6 ILC3 cells increased effector T cells in the intestinal lamina propria, increased inflammatory cytokine secretion, and increased intestinal inflammation (2830)(Goto et al., 2014; Hepworth et al., 2013).These cells present intestinal antigens on MHCII to effector T cells, but instead of inducing cell proliferation, they induce apoptosis (29). In this manner, CCR6 ILC3 MHCII antigen presentation serves a novel immune suppressive function in the intestine.

Commensal antigen-specific responses

After phagocytosing and processing antigen, DCs present antigens to T cells and promote their differentiation. A complex combinations of signals from DCs, intestinal microbes, intestinal epithelial cells, and other immune cells dictate T cell responses, altering susceptibility to pathogens and inflammatory diseases. Commensal microbes induce many of these antigen-specific T cells, and several T cell subsets, such as T helper 17 cells and inducible T regulatory cells, are severely depleted in germ free animals (31, 32). Specific antigen-specific immune interactions are difficult to study due to the complexity of the microbial community. Those that have been described reveal important features of commensal microbes that induce or modulate antigen-specific immune responses.

Th17 cells are particularly important for mucosal immunity, and genetic mutations disrupting Th17 development or function is associated with increased mucosal fungal infections (Okada et al., 2015; Puel et al., 2011) and Th17 deficient mice are more susceptible to intestinal Citrobacter rodentium infection (3335)(Mangan et al., 2006). Th17-defining cytokines, IL-17 and IL-22, promote intestinal barrier function, and induce both antimicrobial peptide and IgA secretion (36). These inflammatory T cell populations also contribute to a number of pathogenic inflammatory conditions, that include inflammatory bowel disease, multiple sclerosis, and arthritis, so their origins are of great interest (37). Intestinal Th17 levels are highly dependent on the microbiota composition, and genetically identical mice from different mouse vendors have can drastically different Th17 levels (31). In this particular case, a single commensal organism, segmented filamentous bacteria (SFB), drove this Th17 response and most SFB-induced Th17 cells are SFB-specific (38). Using TCR hybridomas from SFB-colonized mice, Yang and colleges showed that the majority of the Th17 TCRs specifically recognize SFB antigens (39). Further, SFB Th17 responses require MHCII expression on dendritic cells and specifically require CD103−CD11b+CX3CR1+ DCs (30, 40).

Why does SFB preferentially induce Th17 differentiation? Two recent studies demonstrated that a key feature of SFB required for Th17 induction is its close association with the gut epithelial cells (41, 42). Using mouse and rat-specific SFB strains, Atarashi and colleges found that only strains matching their original host could induce Th17 cells, and this was due to species-specific adherence (41). SFB adherence induced the expression of serum amyloid proteins (SAA1/2), and the reactive oxygen producing protein Duox2 in epithelial cells (38, 41, 42). SAA1, in particular, directly enhanced IL-17A and IL-17F induction by Th17 cells, and also acts on dendritic cells to promote Th17 induction (41, 42).

Regulatory T cells are also induced and regulated by the microbiota and play a vital role in preventing pathogenic inflammation (43). Commensal microbes induce Tregs through number of innate immune and metabolic signaling pathways (discussed later in this review), but do Tregs recognize commensal antigens? To explore this question, two studies took advantage of mouse models with limited but diverse TCR repertoires and compared Treg TCR sequences in different lymphoid tissues (44, 45). These studies demonstrated that intestinal Tregs utilized a distinct set of TCRs compared to other lymphoid tissues and found that these Treg TCR repertoires were significantly altered in germ-free or antibiotic-treated mice. In addition, both groups found that colonic Treg hybridomas specifically recognize diverse commensal bacteria (44, 45). Together, these findings demonstrate that a large portion, if not the majority, of intestinal Tregs recognize commensal antigens.

Mechanisms by which commensal influence specificity of the immune response

Commensals, such as SFB, have been associated with a number of autoimmune diseases. Autoimmune diseases are characterized by inappropriate responses to self antigen and thus it is unclear how commensals might influence these specific immune responses. Here we describe three potential mechanisms that have been highlighted in recent literature, expansion of T cells expressing dual TCRs, bystander activation and molecular mimicry (Figure 2) While GF mice have been shown to be resistant to the development of EAE, mono-association with SFB alone induces worsened disease that was associated with induction of (46) model of arthritis in GF mice and introduction of SFB was sufficient to induce development of Th17 and T follicular helper cells (TFH) to initiate disease (37)(47). Lung pathology is a significant complication and cause of mortality in individuals with rheumatoid arthritis (RA) (48). SFB was shown to exacerbate lung pathology in the K/BxN model of arthritis and this was dependent on Th17 cells (49). SFB was shown to preferentially expand T cells expressing two TCRs, one TCR was specific for SFB and the other for self-antigen. This provides a novel mechanism by which commensal bacteria might influence autoimmunity, through expansion of T cells expressing TCRs against self peptides.

Figure 2:

Figure 2:

How commensals can dictate immune specificity. (A) Organisms within the gut can expand populations of T cells that express two TCRs, one TCR that recognizes the commensal itself and one that reacts against self peptide. Migration of these T cells to host tissues could cause pathology and autoimmunity. (B) Both CD4+ and CD8+ T cells can be activated independently of their TCR. In this scenario, a commensal organism may induce a particular cytokine or be detected directly by T cells through toll-like receptors (TLRs). (C) Commensal organisms might express epitopes that mimic self peptides known to drive autoimmunity. Self reactive T cells would then be activated by these antigens in the gut and subsequently traffic to tissues and elicit autoimmunity.

Another mechanism by which commensals might influence the specificity of the immune response that leads to autoimmunity is through molecular mimicry. Recently, a peptide that is part of a bacterial integrase within the Bacteriodes genes was identified that is identical to a self pancreatic antigen known to elicit inflammation in a mouse model (50). Indeed, this bacterial antigen could be presented by MHCI to stimulate CD8+ T cells. Extending their findings to humans, the authors found the T cells isolated from individuals with type 1 diabetes (T1D) and Crohn’s disease expanded in response to this bacterial antigen (50). Interestingly, T cells specific for this antigen were shown to be protective in a model of colitis, suggesting that these T cells were preserved within the host because they promoted tolerance in the gut. However, this represents an example of how commensal organisms within the gut might express antigens that cause expansion of T cells against other antigens, either self or foreign.

Up to this point we have analyzed the immune responses elicited by the microbiota through a rather classical lens. However, is it really possible that all microbes in the gut activate specific immune responses against themselves? Recent evidence suggests that commensals can also induce responses that are directed against other commensals, what is often referred to as bystander activation (51). Gomes-Neto et. al. recently demonstrated that Helicobacter bilis colonization directs Th17 responses against other members of the microbiota, but not to itself (52). By utilizing the defined ASF microbiota community, this group identified specific microbes targeted by H. bilis-induced Th17 responses. Interestingly, removing Th17-targeted bacteria from the community resulted in Th17 responses against a different collection of microbes, suggesting that H. bilis can alter T cell responses to a broad range of microbes (52). What mechanisms may drive these bystander responses? One possibility is that microbes or treatments that broadly alter intestinal permeability or cytokine milieu. For example, commensal microbes transiently escape the intestine during a Toxoplasma gondii infection and induce long-lived memory Th1 responses (53). Similarly, IBD is associated with Th1 responses specific for commensal flagella proteins in the intestine (54). Innate immune signaling on T cells has been found to induce bystander effects. Stimulation of T cell TLRs by Bacteriodes fragillis promotes Treg differentiation (5557). Recently, the direct TLR stimulation of T cells was found to induce arthritis during a Borrellia infection, the causative agent of Lyme disease (58).

Specificity of intestinal IgA responses

One of the primary functions of activated T cells is to induce and modulate intestinal antibody response. In the gut, the primary secreted immunoglobulin is IgA, which is produced by IgA plasma cells in the lamina propria and transported across the epithelial cell lay via the polymeric Ig receptor (pIgR) (59). IgA plasma cells develop through T cell dependent (TD) and T cell independent (TI) pathways. TI responses develop in the LP or in isolated lymphoid follicles and result from the direct response of B cells to free or DC-delivered antigens (60). Differentiation of these B cells into IgA plasma cells is then induced by multiple signals from DCs and epithelial cells. T cell dependent IgA induced in the PP and requires a specific group of T helper cells, call T follicular helper cells (Tfh). Tfh cells interact with MHCII-presented antigens on B cells in germinal centers (GC) to promote IgA PC development (61). New techniques that combine flow cytometry with 16S sequencing has revealed exciting insights into how IgA interacts with the microbiota, the microbial community that is bound by IgA, and relative role of TD and TI IgA responses in targeting intestinal microbes (summarized in Figure 3) (55, 6265).

Figure 3: TD and TI IgA interactions in the gut.

Figure 3:

T-cell dependent (TD) IgA arises from antigen-specific Tfh/germinal center B cell interactions in the PP and results in highly affinity sIgA. For T cell-independent IgA, naïve B cells can mature into IgA-secreting plasma cells in the PP, isolated lymphoid follicles, or in the lamina propria, resulting in low-affinity, polyreactive sIgA. Most of the IgA coating intestinal microbes comes from TI-IgA secretion. The propotion of IgA-coated bacteria is highest in the SI (40–80%) and decreases in the colon and feces (1–30%). TD-IgA is required for shaping mucosal-associated microbial communities and has been shown to control Ruminococcus, Lachnospiraceae, Desulfobibrionaceae, and Mucispirillum populations. Interestingly, the mucus or tissue associated bacteria, Mucispirillum and SFB are specifically coated by TD-IgA.

Recent work from Bendelac and colleges suggests that most of the IgA bound to commensal is TI-generated. Bunker et al. found that T cell deficient mice had similar levels of IgA coated bacteria throughout their intestine, and the bacterial community bound to IgA was largely similar to WT animals (65). They also demonstrated that IgA interaction is primarily dictated by its location within the intestine; microbes preferentially colonizing the SI are highly coated by IgA, while colon-resident bacteria are not (Figure 3)(65). In a subsequent study, this group demonstrated that IgA plasma cells have a propensity to secrete poly-reactive microbiota-targeting antibodies (66). They cloned and created monoclonal antibodies (mAbs) from sorted IgA plasma cells and compared their specificities to mAbs cloned from naïve B cells. The majority of IgA-derived mAbs bound a broad range of intestinal microbes, while mAbs from naïve B cells did not display this microbiota-reactivity (66). Together, these studies suggest that the majority of the intestinal IgA-bound bacteria are targeted in a non-specific manner. It is still unclear, however, how these TI responses shape the microbiota and alter interactions with the host. Interestingly, microbiota-reactive mAbs primarily bind to bacteria isolated directly from mice, and microbes cultured from this community no longer bound IgA-derived mAbs. This suggests that intestinal microbes either express, or are coated with molecules the bind polyreactive IgA specifically in the gut. Indeed, IgA-derived mAbs bound to a variety of bacterial glycans, raising the interesting possibility that intestinal IgA acts as an innate, perhaps glycan-recognizing, immune response toward microbiota (66). Notably, TD responses appeared to be required for IgA interaction with two mucosal-associated bacteria, SFB and Mucispirillium, and these organisms were not coated by TI IgA (65). Perhaps these organisms have mechanisms to avoid innate IgA interactions and may offer interesting microbes to explore how TI IgA impacts host/microbe interaction.

While TD IgA may not be the primary source of IgA interacting with intestinal microbes, several studies have demonstrated that these responses are important for shaping the microbial community. Disrupting T cells, B cells reduces diversity in the intestinal microbiota (62). To identify specific T cell responses required for maintaining fecal microbiota diversity, Kawamoto et. al., transferred different T cell populations into T cell deficient animals. They found that Foxp3 Tregs as the key T cell population that promotes diversity. Tregs are able to become T follicular regulatory cells in the PP, promoting germinal center formation and boosting IgA plasma cells in the SI. Interestingly, the Treg-shaped microbiota could also boost TD IgA responses when transferred to germ free mice (62). This suggests that there is a positive feedback loop, where TD IgA responses shape a microbiota that further promotes these immune responses. In this study, TD responses promoted Clostridia diversity, likely by suppressing Lachnospiraceae overgrowth (62), and Clostridia species are known to induce Tregs in the gut (32). In addition, our lab demonstrated that disruption of the T cell MyD88, a signaling protein required for the innate immune Toll like receptor signaling, is required for maintaining Tfh cells. Decreased Tfh cell number resulted in significant alterations to the tissue-associated microbial community and exacerbated colitis, which was dependent on changes to the microbiota (55). Therefore, TD induced IgA may not be the primary source of IgA interacting with intestinal microbes but it is important for in shaping the intestinal microbial community and impacting host health (Figure 3).

What is the functional difference between TD and TI IgA? The only documented difference so far is their specificity, with TD IgA having undergone affinity maturation while TI antibodies have not. Intestinal IgA serves multiple protective roles that include direct killing, opsonizing, and agglutination (59), and strength of IgA interaction likely effects each of these functions. For example, IgA prevents Salmonella Typhimurium from fully separating after cell division, preventing spread and promoting clearance. Only vaccine-induced, high-avidity IgA induces chain formation (67). In addition, IgA can directly modulate microbial behavior in the gut and is typically dependent TD IgA. A single specific IgA antibody developed against a Bacteriodes thetaiodomicron capsule epitope can suppress gene transcription required for its production in vivo (68). In addition, mice lacking TLR5, an innate immune receptor detecting bacterial flagella, have reduced flagella-specific IgA and a corresponding increase in microbiome expression of flagella genes (69). These examples suggest that strength and selectivity of IgA responses significantly regulate host/microbe interactions in the gut and these features may differentiate TD and TI responses.

Commensal regulation of host immunity

Just as pathogens have evolved elegant ways to subvert host immunity, commensals have evolved just as intricate mechanisms to control it. Over the last several years, a variety of molecules and metabolites that are produced by the microbiota produced have been identified to directly influence the mammalian immune response. These molecules have been classified by others into 3 specific categories and, here, we introduce a fourth (70). These are 1) specialized secreted microbial molecules 2) microbial modified host molecules 3) metabolism of dietary compounds and 4) and structural microbial components. The most well-known microbial modified host molecules (category 2) are bile acids and their effects on the immune system have been reviewed extensively by others (7072). Therefore, here we will focus on the other 3 categories of microbial compounds.

Specialized secreted molecules

Fecalibacterium prauznitzii is a gut organism that is found in high abundance within the intestine of many individuals (73, 74). Loss of F. prauznitzii is predictive for relapse of Crohn’s disease after surgery and has been demonstrated to protect from the development of colitis in pre-clinical animal models of IBD (73). Recently, a protein termed microbial anti-inflammatory molecule (MAM) was identified in the supernatants of F. prauznitzii cultures(75). Seven peptides were shown to be a part of the F. prauznitzii MAM protein and induce IL-10 in vitro. As the technology to genetically manipulate F. prauznitzii does not exist, the ability to study MAM in detail is difficult. To get around this, the cDNA for this protein was placed into a plasmid and transfected into Lactococcus lactis (76). Animals treated with the MAM expressing L. lactis strain were protected from the induction of colitis using two-independent models. Mechanistically, MAM was shown to inhibit the activation of NF-kB which led to reductions in Th1 and Th17 responses in the gut. Other proteins having similar activity have been isolated from Bifidobacterium animalis subsp. lactis, however are less characterized (77). Thus, loss of F. prauznitzii or Bifidobacterial species within the intestine of individuals, as is seen in patients with Crohn’s disease, would exacerbate inflammation and worsen disease symptoms due to a failure to down-regulate inflammation.

Staphylococcus epidermidis is a common commensal found on skin. While ligation of TLR signaling on keratinocytes elicits potent inflammation, S. epidermidis, despite expression of TLR ligands, does not elicit inflammatory responses from these cells (78). Based on this, it was hypothesized that S. epidermidis expressed a molecule that would antagonize TLR signals. Indeed, a small molecule secreted from S. epidermidis was identified that could selectively antagonize TLR3 signals. Lipoteichoic acid (LTA) is produced in a cellular form that is found within the membrane and an exocellular form that is secreted and can be recovered in a small molecule fraction (<10kDa). Both forms of LTA isolated from S. epidermidis could selectively suppress TLR3 induced inflammation, which is induced, in this study, by wounding the skin. TLR3 suppression mediated by S. epidermidis occurred through TLR2 signaling. Therefore, S. epidermidis is able to limit cutaneous skin inflammation during wound healing. This same group also identified that this molecule was able to elicit anti-microbial peptide expression that could prevent growth of Staphlococcus aureus (79). Since S. epidermidis is a common organism that is associated with sepsis, use of the organism itself as a probiotic would not be advisable (80). However, it is tempting to speculate the use of a highly purified form of S. epidermidis LTA as a naturally occurring anti-inflammatory in a skin cream that might help wounds heal faster and/or prevent growth of pathogenic organisms.

More recently, Powrie and colleagues identified a large polysaccharide found within the supernatants of Helicobacter hepaticus cultures that also induces anti-inflammatory responses(81). H. hepaticus can be a peaceful resident of the commensal microbiota, however if the host is deficient in the anti-inflammatory cytokine, IL-10, H. hepaticus elicits inflammation and causes a chronic colitis (82). H. hepaticus was recently shown to induce IL-10 in gut macrophages without a corresponding increase in pro-inflammatory cytokines. This suggested that H. hepaticus might selectively induce anti-inflammatory responses. Through a series of in vitro experiments, the authors narrowed down a secreted polysaccharide from H. hepaticus cultures as being responsible for induction of IL-10. Interestingly, induction of IL-10 by this polysaccharide was dependent on signaling through TLR2 and MyD88 as is seen with B. fragilis and S. epidermidis (56).

While these molecules seem to be specific to these organisms, other common microbial products, such as ATP, can also direct host immunity. Over a decade ago, it was reported that ATP from commensal bacteria could elicit Th17 responses within the gut (83). Indeed, fecal ATP levels in GF mice were significantly reduced when compared to SPF mice and administration of ATP to GF mice was sufficient to induce Th17 responses within the gut. ATP was sensed by P2X receptors on a specific subset of CD70highCDllclow dendritic cells to induce this inflammatory pathway. More recent studies have shown a role for bacteria derived ATP for induction of TFH cells. P2rx7 is a receptor that binds to ATP and disruptions to this receptor cause weight gain in animals (84). P2rx7−/− TFH cells were sufficient to recapitulate this phenotype, suggesting that TFH cells are directly responsive to ATP (85). To prove that TFH cells were influenced by microbially-produced ATP the authors performed an elegant experiment in germfree mice. They constructed a mutant of E. coli that expressed an apyrase so that all ATP produced by the microbe would be hydrolyzed and no extracellular ATP could be generated. GF animals were colonized with a control E. coli or the apyrase expressing E. coli and TFH cells and IgA were monitored. Animals that were colonized with ATP deficient bacteria had increased TFH and E. coli specific IgA responses compared to controls. These data suggest that ATP produced by commensals can serve to limit host antibody responses that functionally impact the composition of the microbiota leading to metabolic defects. It is likely that a multitude of organisms secrete ATP, therefore it is unclear how ATP would differentially drive IL-17 versus TFH responses within gut. Perhaps the location of the ATP, or the specific cell type that is detecting the ATP would drive these differential outcomes (86).

In addition to detection of bacterially produced molecules through receptor mediated interactions, microbes are also known to make proteases that can directly destroy host proteins such as IgA (59). Lactobacillus paracasei has beneficial effects within the mammalian gut and is one of the members of a probiotic termed VSL#3 (87). Feeding animals VSL#3 during colitis, significantly downregulates inflammation and ameliorates colitis symptoms (88). This activity was shown to be reliant in part on a molecule that was secreted by L. paracasei. Further analysis identified this molecule as a serine protease that can degrade an array of pro-inflammatory chemokines such as IP-10 (89, 90). Disruption of this bacterial protease in the organism lead to a defect in IP-10 degradation and a failure to protect from colitis. Similarly, a commensal yeast strain known to ameliorate colitis induction in animals, Saccharomyces boulardii, was reported to secrete a protease that degrades toxin A produced by Clostridium difficle and protects from enteritis(91). Thus, commensal microbes express and secrete molecules that can actively degrade inflammatory cytokines as a mechanism to maintain tolerance within the intestine.

Dietary metabolites

There are several metabolites that arise from microbial metabolism of dietary compounds. These include, short-chain fatty acids (SCFAs), amino acid metabolites such as tryptophan and arginine, flavonoids and polyamines. SCFAs, include propionate, butyrate and acetate and are a result of fermentation by the microbiota. These metabolites have been demonstrated to induce Treg responses and thus down-regulate inflammation (32, 92). SCFAs also serve as a source of energy for intestinal epithelial cells and influence barrier function (93, 94). These particular metabolites have been extensively reviewed elsewhere (70, 72) and therefore will not be covered in depth within this review.

Tryptophan metabolites

Changes to microbial derived tryptophan metabolites are beginning to emerge in a number of human diseases (9598). The microbiota metabolizes dietary tryptophan to indole as well as multiple indole derivatives including indole-3-acetate, indole-3-aldehyde, indole-proprionic acid (IPA), indole acetic acid, indoxyl-3-sulfate, and indole acrylic acid. Multiple organisms have been shown to metabolize tryptophan including E. coli, Lactobacillus species and Clostridia species such Clostridia sporogenes.

Indole itself was demonstrated to influence intestinal epithelial cell gene expression patterns, mucin production and enhanced tight junctions. Indole, indole-3-acetate, indole-3-aldehyde and tryptoamine have all been identified to act on the aryl-hydrocarbon receptor (AHR)(99). AHR activation can lead to expansion of innate lymphoid cells and IL-22 production leading to the development of intestinal lymphoid follicles (100102). Mammals produce the enzyme indoleamine 2,3,-dioxygenase (IDO) to degrade tryptophan. To investigate host-extrinsic mechanisms of tryptophan degradation, Romani and colleagues utilized IDO−/− mice, which increases tryptophan availability to gut microbes(103). In these animals, Lactobacilli become expanded and produce copious amounts of indole-3-aldehyde. This metabolite directly upregulated expression of IL-22 through AHR and provided colonization resistance from C. albicans infection (103).

Polymorphisms in Card9 are found in individuals with IBD (104). Card9 is a signaling molecule that lies downstream of Dectin-1 and is activated by fungal cell wall components. Card9−/−animals are more susceptible to colitis and this is dependent on the microbiota, as transfer to a GF recipient causes worsened colitis (95). The microbiota formed within these animals displayed the inability to metabolize tryptophan. In particular, indole-3-acetic acid was significantly lower in Card9−/− animals. As these tryptophan metabolites are known to bind AHR, the authors administered a known AHR agonist to Card9−/− and this ameliorated colitis (95). This studies suggests that loss of specific microbial metabolites can exacerbate disease.

Tryptophan metabolites have also been shown to function at extra-intestinal sites such as the central nervous system(96). IFN-β was shown to limit inflammation associated with a pre-clinical animal model of multiple sclerosis and its activity was in part dependent on AHR expression within astrocytes. A diet lacking tryptophan worsened disease, while replacement of the microbial metabolite, indole-3 sulfate, ameliorated disease. Indole-propionic acid and indole-3-aldehyde had similar protective effects. To directly demonstrate that gut bacterial metabolism can influence extra-intestinal immune responses, Sonnenburg and colleagues identified the gene cluster in C. sporogenes responsible for metabolizing tryptophan to indole-propionic acid. Colonization of animals that lack this gene cluster fail to generate IPA and this causes enhanced T cell activity and antibody circulation within the blood(105). IPA is a known agonist for the pregnane X receptor (PXR) and thus the mechanism of action of this metabolite might be PXR dependent as has been shown within the gut (106). Thus, intestinal microbial metabolites can get into circulation where they bind to AHR and limit CNS inflammation.

Mucus serves as an energy source for multiple microbes within the gut and mucin-degraders can have both beneficial and detrimental consequences to the host (107, 108). To identify organisms that degrade mucins Xavier and colleagues utilized a computational approach (98). This screen lead to multiple organisms within the Clostridales order and 36 of them were able to grow on mucin. Further testing identified strains that were protective during DSS colitis, one of which was Peptostroptococcus russellii. Genome sequencing of this organism identified that it contained the fldA/BC loci known to be responsible for producing tryptophan metabolites. P. russellii made indoleacrylic acid which prevented intestinal permeability and down-regulated inflammatory responses.

Biogenic amines

Microbial metabolism of amino acids can produce biogenic amines through de-carboxylation. These include cadaverine (from lysine), agamatine, spermine and spermidine (from arginine) and histamine (from histidine). Most of these amines can also be made by the host and therefore, the literature on microbiota derived biogenic amines and there influence on the host is still in its infancy(109). Many of these biogenic amines however are known to have important consequences to the host. Purified agmatine administered to rats after the induction of cerebral ischemia significantly reduced brain infarction and minimized neuro-inflammation (110). Similarly, agmatine treatment attenuated fever associated with LPS administration (111) and its metabolite, spermine, protected mice from lethal sepsis and had suppressive immune effects (112, 113). With the known contribution of the microbiota to CNS health and development, this might be but one mechanism by which the microbiota can influence CNS health that has yet to be explored.

Histamine is another biogenic amine that has recently been shown to have beneficial effects on the immune system. Purified histamine cultured with intestinal explants suppresses IL-18 induction, demonstrating an anti-inflammatory role for this metabolite (113). Other studies identified a histamine secreting microbe could down-regulate inflammation. There are four G-protein coupled receptors encoded within humans that recognize histamine. Loss of the histamine receptor 2(HR2) in animals leads to the activation of multiple pro-inflammatory cytokines within the gut. A histamine secreting microbe, Lactobacillus rhamnosus, suppresses the production of TNF-α and GM-CSF within the gut in a HR2 dependent manner (114). Thus, metabolic conversion of dietary amino acids by the microbiota can have pervasive effects on the host immune system.

While most of the functional metabolites described above provide some benefit to the host, not all microbial metabolites fall into this category. Choline is a quaternary amine required for multiple biological processes in mammals. Metabolism of choline by the gut microbiota results in the production of trimethylamine (TMA)(115). TMA is absorbed by the intestine and can further be metabolized in the liver into trimethylamine-N-oxide (TMAO). TMAO levels have been positively correlated with several diseases including diabetes, atherosclerosis, non-alcoholic fatty liver disease and impaired renal function (116118). TMAO injection into animals causes activation of NF-κB and elicits inflammation with the aorta. Additionally, microbes that utilize dietary choline within the gut, limit its bioavailability to the host (119). This could be especially harmful during pregnancy, where the fetuses need for dietary choline is significantly high. To test this, Balskus and collegues colonized dams with high levels of choline-consuming microbes which produced offspring with behavioral alterations. Thus, microbes can also compete for host nutrients to the host detriment.

Molecules that are part of the microbe structure

As an immune defense strategy, recognition of molecules exclusively expressed by micro-organisms is an elegant mechanism of sensing invasion and eradicating potential infection. Immune responses elicited upon detection of foreign bacterial molecules by host immune systems has been studied extensively and served as the basis for the many of the paradigms that have been identified to differentiate between ‘self’ and ‘non-self’. However, many of these same foreign molecules that are known to trigger sterilizing immunity are also expressed by commensal organisms and are now known to prime the development of steady state immune responses. This has led to speculation that perhaps many of these receptors evolved, at least in part, to initiate tolerance against commensal microbes(120, 121).

Some of the most commonly studied structural bacterial molecules are peptidoglycan (PGN), lipopolysaccharide (LPS) and flagellin. PGN can be recognized by a number of receptors including TLR2, NOD1, NOD2, NLRP3 through hexokinase (122), and PGN recognition proteins (PGRPs)(123) while LPS and flagellin are known to stimulate TLR4 and TLR5 respectively. These commensally derived molecules are found in abundance within the gut, but can also be found circulating within the blood where they have been shown to prime immunity against pathogenic infection and in some cases, induce tolerant immune responses against commensals.

PGN is recognized by a number of host receptors and polymorphisms within many of these genes, including TLR2 and Nod2, are associated with IBD in humans (124126). Disruption of any one of these receptors in mice leads to increased susceptibility to colitis in multiple studies. However, in many cases, these results are contentious and either depend on the type of colitis model used or the facility in which the experiments were performed. As an example, TLR2 deficient animals are more susceptible to damage associated models of colitis induced by the chemical DSS, and provision of a synthetic TLR2 agonist to wild-type mice can ameliorate mucosal inflammation in this same model. However, in models of more chronic intestinal inflammation, such as the T cell transfer model of colitis, TLR2 does not have an effect on the development of disease (127). These disparate results could be explained by the different models used, as one is mediated by epithelial damage and the other is mediated by chronic T cell inflammation. However, changes in the microbiota composition from facility to facility might also result in these differences (128). As colitis results, in part, from a loss of tolerance to commensal bacteria, the composition of the microbiota is imperative to the severity of disease. Indeed, certain microbial compositions are sufficient to worsen colitis while others are protective. A common theme amongst animals that lack PGN recognition, is that the composition of the microbiota was significantly different when compared to wild-type animals. This is true of TLR2−/− animals (129), NOD2−/−(128, 130, 131), and PGRP1–4−/−(123) animals. Similarly, a deficiency in receptors recognizing flagellin (132) and LPS (133) have reported changes to the structure of the microbiota in mutant mice. While differences in microbial composition due to mutations within host immunity is more of a rule, rather than an exception; changes in composition are not always required for exacerbated disease development. Thus, it is important to perform co-housing, germfree transplants, and/or antibiotic disruptions to demonstrate causality of the microbiota. This has been done in experiments using TLR5 (132) and PGRP1–4(123) deficient animals. To prove that the composition of the microbiota was sufficient to influence disease severity, the microbiota from mutant mice was transplanted into wild-type GF recipients. GF mice receiving the transplant from mutant animals developed worsened disease than GF mice receiving a transplant from the healthy animals. As the microbiota differs in various animal facilities, this may provide an explanation for disparate results found in the same mutant mice from different institutions.

While commensal cell wall components seem to be important in dictating microbial composition within the gut, commensal cell wall products have also been detected in circulation. Here they are reported to directly prime immune responses and protect from infection associated damage. Animals treated with antibiotics have a significantly increased susceptibility to influenza that includes delayed viral clearance and enhanced bronchiole damage (134). Loss of commensal bacteria upon antibiotic treatment impaired the induction of anti-viral immunity within peritoneal macrophages. Viral infection is in part sensed through dsRNA binding to TLR3. Remarkably, intra-nasal administration of synthetic dsRNA (poly I:C) to antibiotic treated animals restored their ability to mount anti-viral immunity and clear viral infection. Similarly, PGN was identified within circulating blood and shown to prime neutrophils within the bone marrow (135). GF mice have undetectable levels of PGN within blood and have increased susceptibility to Streptococcus pnenmonia and Staphlococcus aerus. Oral administration of PGN to GF mice is sufficient to restore neutrophil function and protect from infection. These results demonstrate that commensal bacteria provide tonic signaling that can prime extra-intestinal immune responses.

Sphingolipids are a distinctive group of membrane lipids that are made up of long-chain, monounsaturated, di-hydroxy amine structures. These lipids are critical components of the plasma membrane in mammals, but are also found on certain bacterial and fungal organisms. Bacterial sphingolipids are predominantly produced by organisms within the phylum Bacteriodetes, but can also be found in the fungal cell walls of Saccharomyces and Candida species (136). Interestingly, invariant natural killer T cells (iNKT) recognize nonpolymorphic CD1d glycosphingolipids, demonstrating recognition of these molecules by host immune systems (137). Bacteriodetes are largely found associated with mammalian hosts and therefore one group sought to determine whether bacterial produced sphinolipids influenced host immune system development. They identified a gene encoded in Bacteroides fragilis that possessed significant homology with the eukaryotic enzyme serine palmitoyltransferase (SPT)(138), which is the first enzyme involved in sphingolipid biosynthesis. Germfree mice have increased iNKT cells within the colonic lamina propria, mono-associations of GF animals with B. fragilis could decrease this population to the same levels as specific pathogen free (SPF) mice. Deletion of the enzyme involved in sphingolipid synthesis in B. fragilis failed to reduce the population of iNKT found in GF mice, indicating that bacterial sphingolipids negatively regulation host iNKT populations. An overabundance of iNKT cells within the gut could be detrimental to the host during intestinal disease. Treatment of animals with the purified B. fragilis sphingolipid protected animals from the development of colitis, indicating a beneficial role for bacterial sphingolipids.

Polysaccharide A (PSA) is one of eight polysaccharides that are found in the capsule of the prominent commensal species, B. fragilis. PSA is part of the capsule of B. fragilis but it also secreted in outer membrane vesicles, where it can be delivered to host cells. PSA has been shown in multiple studies to act as an agonist for TLR2 and induce tolerogenic responses (56, 139, 140). The use of purified PSA can protect animals from the development of intestinal colitis and multiple sclerosis (140, 141). B. fragilis is known to associate closely within intestinal crypts and disruption of PSA in B. fragilis fails to induce host Tregs and B. fragilis can no longer associate with the intestinal epithelium (56). The effects of PSA have been discussed elsewhere(70), but the many studies on this one commensal molecule serve as an example of the powerful therapeutic potential that can be uncovered through the detailed study of commensal organisms.

While less abundant within the gut, fungal species are indeed relevant during intestinal development and disease. Multiple polymorphisms within genes that recognize fungal cell wall components have been found in individuals with IBD, including Dectin-1 and Card9 (142). However, Nod2 is one of the most commonly found mutations within individuals with Crohn’s (143). It is known to be stimulated by PGN and most studies to date have focused on Nod2 recognition of PGN. However, Nod2 has also been reported to be stimulated by fungal chitin, an essential cell wall component found within most fungal organisms (144). Stimulation through NOD2 with chitin elicits anti-inflammatory IL-10 production, suggesting that tonic signaling through Nod2 by fungal organisms can elicit tolerogenic responses within the gut. Fungal cell wall mannan is also highly immune stimulatory as is the target of multiple C-type lectin and TLR innate immune receptors (145). Recently, fungal mannan was sufficient to rescue exacerbated colitis and increased susceptibility toward influenza in antibiotic-treated mice, in a process that required TLR4 (146). Fungal organisms can also express specialized polysaccharides that can influence host responses. Although rarely discussed within the context of commensalism, Cryptococcus neoformans, also expresses an immune-suppressive polysaccharide capsule comprised of glucuronoxylomannan (GXM) and galactoxylomannan (GalXM). Both GXM and GalXM have multiple anti-inglammatory properties and include inhibiting TLR4 signaling, promoting macrophage secretion of FasL to promote T cell apoptosis (147). While C. neoformans is widely studied as a pathogen that causes high mortality in immune-compromised patients, it rarely induces disease in healthy individuals. C. neoformans prevalent environment and likely encountered often by individuals and therefore might influence steady state responses in the host. Indeed, the use of GXM was recently found to be therapeutically useful in models of sepsis (148). Thus, eukaryotic species residing at mucosal surfaces can also directly impact host immunity.

While much work has been done already to decipher how gut microbes influence their mammalian hosts, much remains to be done. With hundrends of distinct organisms from various kingdoms of life, little is known about the interactions that occur between them and consequences to the disease state. Given the importance of these microbes to human health it is likely that treatments to many diseases might be uncovered within our own GI tract.

REFERENCES

  • 1.Honda K, Littman DR. 2016. The microbiota in adaptive immune homeostasis and disease. Nature 535: 75–84 [DOI] [PubMed] [Google Scholar]
  • 2.Thaiss CA, Zmora N, Levy M, Elinav E. 2016. The microbiome and innate immunity. Nature 535: 65–74 [DOI] [PubMed] [Google Scholar]
  • 3.Bolnick DI, Snowberg LK, Caporaso JG, Lauber C, Knight R, Stutz WE. 2014. Major Histocompatibility Complex class IIb polymorphism influences gut microbiota composition and diversity. Mol Ecol 23: 4831–45 [DOI] [PubMed] [Google Scholar]
  • 4.Kubinak JL, Stephens WZ, Soto R, Petersen C, Chiaro T, Gogokhia L, Bell R, Ajami NJ, Petrosino JF, Morrison L, Potts WK, Jensen PE, O’Connell RM, Round JL. 2015. MHC variation sculpts individualized microbial communities that control susceptibility to enteric infection. Nat Commun 6: 8642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rosenbaum JT, Davey MP. 2011. Time for a gut check: evidence for the hypothesis that HLA-B27 predisposes to ankylosing spondylitis by altering the microbiome. Arthritis Rheum 63: 3195–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Silverman M, Kua L, Tanca A, Pala M, Palomba A, Tanes C, Bittinger K, Uzzau S, Benoist C, Mathis D. 2017. Protective major histocompatibility complex allele prevents type 1 diabetes by shaping the intestinal microbiota early in ontogeny. Proc Natl Acad Sci U S A 114: 9671–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Toivanen P, Vaahtovuo J, Eerola E. 2001. Influence of major histocompatibility complex on bacterial composition of fecal flora. Infect Immun 69: 2372–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Noble JA, Valdes AM. 2011. Genetics of the HLA region in the prediction of type 1 diabetes. Curr Diab Rep 11: 533–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mathis DJ, Benoist C, Williams VE 2nd,, Kanter M, McDevitt HO. 1983. Several mechanisms can account for defective E alpha gene expression in different mouse haplotypes. Proc Natl Acad Sci U S A 80: 273–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bogunovic M, Mortha A, Muller PA, Merad M. 2012. Mononuclear phagocyte diversity in the intestine. Immunol Res 54: 37–49 [DOI] [PubMed] [Google Scholar]
  • 11.Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. 2007. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol 8: 1086–94 [DOI] [PubMed] [Google Scholar]
  • 12.Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M. 2009. Origin of the lamina propria dendritic cell network. Immunity 31: 513–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, Pabst O. 2009. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med 206: 3101–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. 2012. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature 483: 345–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Johansson-Lindbom B, Svensson M, Pabst O, Palmqvist C, Marquez G, Forster R, Agace WW. 2005. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J Exp Med 202: 1063–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Macpherson AJ, Uhr T. 2004. Induction of protective IgA by intestinal dendritic cells carrying commensal bacteria. Science 303: 1662–5 [DOI] [PubMed] [Google Scholar]
  • 17.Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR. 2000. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 20: 4106–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. 2009. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity 31: 502–12 [DOI] [PubMed] [Google Scholar]
  • 19.Niess JH, Brand S, Gu X, Landsman L, Jung S, McCormick BA, Vyas JM, Boes M, Ploegh HL, Fox JG, Littman DR, Reinecker HC. 2005. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307: 254–8 [DOI] [PubMed] [Google Scholar]
  • 20.Niess JH, Adler G. 2010. Enteric flora expands gut lamina propria CX3CR1+ dendritic cells supporting inflammatory immune responses under normal and inflammatory conditions. J Immunol 184: 2026–37 [DOI] [PubMed] [Google Scholar]
  • 21.Diehl GE, Longman RS, Zhang JX, Breart B, Galan C, Cuesta A, Schwab SR, Littman DR. 2013. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX(3)CR1(hi) cells. Nature 494: 116–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mazzini E, Massimiliano L, Penna G, Rescigno M. 2014. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1(+) macrophages to CD103(+) dendritic cells. Immunity 40: 248–61 [DOI] [PubMed] [Google Scholar]
  • 23.Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. 2007. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med 204: 1757–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liang J, Huang HI, Benzatti FP, Karlsson AB, Zhang JJ, Youssef N, Ma A, Hale LP, Hammer GE. 2016. Inflammatory Th1 and Th17 in the Intestine Are Each Driven by Functionally Specialized Dendritic Cells with Distinct Requirements for MyD88. Cell Rep 17: 1330–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Luda KM, Joeris T, Persson EK, Rivollier A, Demiri M, Sitnik KM, Pool L, Holm JB, Melo-Gonzalez F, Richter L, Lambrecht BN, Kristiansen K, Travis MA, Svensson-Frej M, Kotarsky K, Agace WW. 2016. IRF8 Transcription-Factor-Dependent Classical Dendritic Cells Are Essential for Intestinal T Cell Homeostasis. Immunity 44: 860–74 [DOI] [PubMed] [Google Scholar]
  • 26.Persson EK, Uronen-Hansson H, Semmrich M, Rivollier A, Hagerbrand K, Marsal J, Gudjonsson S, Hakansson U, Reizis B, Kotarsky K, Agace WW. 2013. IRF4 transcription-factor-dependent CD103(+)CD11b(+) dendritic cells drive mucosal T helper 17 cell differentiation. Immunity 38: 958–69 [DOI] [PubMed] [Google Scholar]
  • 27.Sonnenberg GF, Artis D. 2015. Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat Med 21: 698–708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hepworth MR, Monticelli LA, Fung TC, Ziegler CG, Grunberg S, Sinha R, Mantegazza AR, Ma HL, Crawford A, Angelosanto JM, Wherry EJ, Koni PA, Bushman FD, Elson CO, Eberl G, Artis D, Sonnenberg GF. 2013. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 498: 113–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, Withers DR, Hugues S, Farrar MA, Reith W, Eberl G, Baldassano RN, Laufer TM, Elson CO, Sonnenberg GF. 2015. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4(+) T cells. Science 348: 1031–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Goto Y, Panea C, Nakato G, Cebula A, Lee C, Diez MG, Laufer TM, Ignatowicz L, Ivanov II. 2014. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity 40: 594–607 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ivanov II, Frutos Rde L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, Littman DR. 2008. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4: 337–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY. 2013. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504: 451–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, Alzahrani M, Al-Muhsen S, Halwani R, Ma CS, Wong N, Soudais C, Henderson LA, Marzouqa H, Shamma J, Gonzalez M, Martinez-Barricarte R, Okada C, Avery DT, Latorre D, Deswarte C, Jabot-Hanin F, Torrado E, Fountain J, Belkadi A, Itan Y, Boisson B, Migaud M, Arlehamn CSL, Sette A, Breton S, McCluskey J, Rossjohn J, de Villartay JP, Moshous D, Hambleton S, Latour S, Arkwright PD, Picard C, Lantz O, Engelhard D, Kobayashi M, Abel L, Cooper AM, Notarangelo LD, Boisson-Dupuis S, Puel A, Sallusto F, Bustamante J, Tangye SG, Casanova JL. 2015. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science 349: 606–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L, Lim HK, Migaud M, Israel L, Chrabieh M, Audry M, Gumbleton M, Toulon A, Bodemer C, El-Baghdadi J, Whitters M, Paradis T, Brooks J, Collins M, Wolfman NM, Al-Muhsen S, Galicchio M, Abel L, Picard C, Casanova JL. 2011. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 332: 65–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. 2006. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 441: 231–4 [DOI] [PubMed] [Google Scholar]
  • 36.Schnupf P, Gaboriau-Routhiau V, Sansonetti PJ, Cerf-Bensussan N. 2017. Segmented filamentous bacteria, Th17 inducers and helpers in a hostile world. Curr Opin Microbiol 35: 100–9 [DOI] [PubMed] [Google Scholar]
  • 37.Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D. 2010. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32: 815–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR. 2009. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139: 485–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Yang Y, Torchinsky MB, Gobert M, Xiong H, Xu M, Linehan JL, Alonzo F, Ng C, Chen A, Lin X, Sczesnak A, Liao JJ, Torres VJ, Jenkins MK, Lafaille JJ, Littman DR. 2014. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature 510: 152–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Panea C, Farkas AM, Goto Y, Abdollahi-Roodsaz S, Lee C, Koscso B, Gowda K, Hohl TM, Bogunovic M, Ivanov II. 2015. Intestinal Monocyte-Derived Macrophages Control Commensal-Specific Th17 Responses. Cell Rep 12: 1314–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, Suda W, Imaoka A, Setoyama H, Nagamori T, Ishikawa E, Shima T, Hara T, Kado S, Jinnohara T, Ohno H, Kondo T, Toyooka K, Watanabe E, Yokoyama S, Tokoro S, Mori H, Noguchi Y, Morita H, Ivanov II, Sugiyama T, Nunez G, Camp JG, Hattori M, Umesaki Y, Honda K. 2015. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 163: 367–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, Lee JY, Ziel JW, Miraldi ER, Domingos AI, Bonneau R, Littman DR. 2015. An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses. Cell 163: 381–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Geuking MB, Cahenzli J, Lawson MA, Ng DC, Slack E, Hapfelmeier S, McCoy KD, Macpherson AJ. 2011. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34: 794–806 [DOI] [PubMed] [Google Scholar]
  • 44.Cebula A, Seweryn M, Rempala GA, Pabla SS, McIndoe RA, Denning TL, Bry L, Kraj P, Kisielow P, Ignatowicz L. 2013. Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota. Nature 497: 258–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, Peterson DA, Stappenbeck TS, Hsieh CS. 2011. Peripheral education of the immune system by colonic commensal microbiota. Nature 478: 250–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. 2011. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci U S A 108 Suppl 1: 4615–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Teng F, Klinger CN, Felix KM, Bradley CP, Wu E, Tran NL, Umesaki Y, Wu HJ. 2016. Gut Microbiota Drive Autoimmune Arthritis by Promoting Differentiation and Migration of Peyer’s Patch T Follicular Helper Cells. Immunity 44: 875–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Raghavendran K, Nemzek J, Napolitano LM, Knight PR. 2011. Aspiration-induced lung injury. Crit Care Med 39: 818–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bradley CP, Teng F, Felix KM, Sano T, Naskar D, Block KE, Huang H, Knox KS, Littman DR, Wu HJ. 2017. Segmented Filamentous Bacteria Provoke Lung Autoimmunity by Inducing Gut-Lung Axis Th17 Cells Expressing Dual TCRs. Cell Host Microbe 22: 697–704 e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hebbandi Nanjundappa R, Ronchi F, Wang J, Clemente-Casares X, Yamanouchi J, Sokke Umeshappa C, Yang Y, Blanco J, Bassolas-Molina H, Salas A, Khan H, Slattery RM, Wyss M, Mooser C, Macpherson AJ, Sycuro LK, Serra P, McKay DM, McCoy KD, Santamaria P. 2017. A Gut Microbial Mimic that Hijacks Diabetogenic Autoreactivity to Suppress Colitis. Cell 171: 655–67 e17 [DOI] [PubMed] [Google Scholar]
  • 51.Ruff WE, Kriegel MA. 2015. Autoimmune host-microbiota interactions at barrier sites and beyond. Trends Mol Med 21: 233–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gomes-Neto JC, Kittana H, Mantz S, Segura Munoz RR, Schmaltz RJ, Bindels LB, Clarke J, Hostetter JM, Benson AK, Walter J, Ramer-Tait AE. 2017. A gut pathobiont synergizes with the microbiota to instigate inflammatory disease marked by immunoreactivity against other symbionts but not itself. Scientific Reports 7: 17707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hand TW, Dos Santos LM, Bouladoux N, Molloy MJ, Pagan AJ, Pepper M, Maynard CL, Elson CO 3rd,, Belkaid Y. 2012. Acute gastrointestinal infection induces long-lived microbiota-specific T cell responses. Science 337: 1553–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lodes MJ, Cong Y, Elson CO, Mohamath R, Landers CJ, Targan SR, Fort M, Hershberg RM. 2004. Bacterial flagellin is a dominant antigen in Crohn disease. Journal of Clinical Investigation 113: 1296–306 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kubinak JL, Petersen C, Stephens WZ, Soto R, Bake E, O’Connell RM, Round JL. 2015. MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health. Cell Host Microbe 17: 153–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK. 2011. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332: 974–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Round JL, Mazmanian SK. 2010. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 107: 12204–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Whiteside SK, Snook JP, Ma Y, Sonderegger FL, Fisher C, Petersen C, Zachary JF, Round JL, Williams MA, Weis JJ. 2018. IL-10 Deficiency Reveals a Role for TLR2-Dependent Bystander Activation of T Cells in Lyme Arthritis. J Immunol [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kubinak JL, Round JL. 2016. Do antibodies select a healthy microbiota? Nat Rev Immunol 16: 767–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pabst O. 2012. New concepts in the generation and functions of IgA. Nat Rev Immunol 12: 821–32 [DOI] [PubMed] [Google Scholar]
  • 61.Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K. 2010. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu Rev Immunol 28: 243–73 [DOI] [PubMed] [Google Scholar]
  • 62.Kawamoto S, Maruya M, Kato LM, Suda W, Atarashi K, Doi Y, Tsutsui Y, Qin H, Honda K, Okada T, Hattori M, Fagarasan S. 2014. Foxp3(+) T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 41: 152–65 [DOI] [PubMed] [Google Scholar]
  • 63.Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, Degnan PH, Hu J, Peter I, Zhang W, Ruggiero E, Cho JH, Goodman AL, Flavell RA. 2014. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158: 1000–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Planer JD, Peng Y, Kau AL, Blanton LV, Ndao IM, Tarr PI, Warner BB, Gordon JI. 2016. Development of the gut microbiota and mucosal IgA responses in twins and gnotobiotic mice. Nature 534: 263–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bunker JJ, Flynn TM, Koval JC, Shaw DG, Meisel M, McDonald BD, Ishizuka IE, Dent AL, Wilson PC, Jabri B, Antonopoulos DA, Bendelac A. 2015. Innate and Adaptive Humoral Responses Coat Distinct Commensal Bacteria with Immunoglobulin A. Immunity 43: 541–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bunker JJ, Erickson SA, Flynn TM, Henry C, Koval JC, Meisel M, Jabri B, Antonopoulos DA, Wilson PC, Bendelac A. 2017. Natural polyreactive IgA antibodies coat the intestinal microbiota. Science 358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Moor K, Diard M, Sellin ME, Felmy B, Wotzka SY, Toska A, Bakkeren E, Arnoldini M, Bansept F, Co AD, Voller T, Minola A, Fernandez-Rodriguez B, Agatic G, Barbieri S, Piccoli L, Casiraghi C, Corti D, Lanzavecchia A, Regoes RR, Loverdo C, Stocker R, Brumley DR, Hardt WD, Slack E. 2017. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 544: 498–502 [DOI] [PubMed] [Google Scholar]
  • 68.Peterson DA, McNulty NP, Guruge JL, Gordon JI. 2007. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2: 328–39 [DOI] [PubMed] [Google Scholar]
  • 69.Cullender TC, Chassaing B, Janzon A, Kumar K, Muller CE, Werner JJ, Angenent LT, Bell ME, Hay AG, Peterson DA, Walter J, Vijay-Kumar M, Gewirtz AT, Ley RE. 2013. Innate and adaptive immunity interact to quench microbiome flagellar motility in the gut. Cell Host Microbe 14: 571–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Postler TS, Ghosh S. 2017. Understanding the Holobiont: How Microbial Metabolites Affect Human Health and Shape the Immune System. Cell Metab 26: 110–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Abt MC, Artis D. 2009. The intestinal microbiota in health and disease: the influence of microbial products on immune cell homeostasis. Curr Opin Gastroenterol 25: 496–502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sharon G, Garg N, Debelius J, Knight R, Dorrestein PC, Mazmanian SK. 2014. Specialized metabolites from the microbiome in health and disease. Cell Metab 20: 719–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, Cosnes J, Corthier G, Marteau P, Dore J. 2009. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflammatory bowel diseases 15: 1183–9 [DOI] [PubMed] [Google Scholar]
  • 74.Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G, Thomas G, Blottiere HM, Dore J, Marteau P, Seksik P, Langella P. 2008. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proceedings of the National Academy of Sciences of the United States of America 105: 16731–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Quevrain E, Maubert MA, Michon C, Chain F, Marquant R, Tailhades J, Miquel S, Carlier L, Bermudez-Humaran LG, Pigneur B, Lequin O, Kharrat P, Thomas G, Rainteau D, Aubry C, Breyner N, Afonso C, Lavielle S, Grill JP, Chassaing G, Chatel JM, Trugnan G, Xavier R, Langella P, Sokol H, Seksik P. 2016. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 65: 415–25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Breyner NM, Michon C, de Sousa CS, Vilas Boas PB, Chain F, Azevedo VA, Langella P, Chatel JM. 2017. Microbial Anti-Inflammatory Molecule (MAM) from Faecalibacterium prausnitzii Shows a Protective Effect on DNBS and DSS-Induced Colitis Model in Mice through Inhibition of NF-kappaB Pathway. Front Microbiol 8: 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wang Z, Wang J, Cheng Y, Liu X, Huang Y. 2011. Secreted factors from Bifidobacterium animalis subsp. lactis inhibit NF-kappaB-mediated interleukin-8 gene expression in Caco-2 cells. Appl Environ Microbiol 77: 8171–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, Yang Y, Cogen AL, Wu ZR, Hooper LV, Schmidt RR, von Aulock S, Radek KA, Huang CM, Ryan AF, Gallo RL. 2009. Commensal bacteria regulate Toll-like receptor 3-dependent inflammation after skin injury. Nature medicine 15: 1377–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, Shafiq F, Kotol PF, Bouslimani A, Melnik AV, Latif H, Kim JN, Lockhart A, Artis K, David G, Taylor P, Streib J, Dorrestein PC, Grier A, Gill SR, Zengler K, Hata TR, Leung DY, Gallo RL. 2017. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci Transl Med 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Otto M. 2017. Staphylococcus epidermidis: a major player in bacterial sepsis? Future Microbiol 12: 1031–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Danne C, Ryzhakov G, Martinez-Lopez M, Ilott NE, Franchini F, Cuskin F, Lowe EC, Bullers SJ, Arthur JSC, Powrie F. 2017. A Large Polysaccharide Produced by Helicobacter hepaticus Induces an Anti-inflammatory Gene Signature in Macrophages. Cell Host Microbe 22: 733–45 e5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kullberg MC, Ward JM, Gorelick PL, Caspar P, Hieny S, Cheever A, Jankovic D, Sher A. 1998. Helicobacter hepaticus triggers colitis in specific-pathogen-free interleukin-10 (IL-10)-deficient mice through an IL-12-and gamma interferon-dependent mechanism. Infection and immunity 66: 5157–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, Takeda K. 2008. ATP drives lamina propria T(H)17 cell differentiation. Nature 455: 808–12 [DOI] [PubMed] [Google Scholar]
  • 84.Beaucage KL, Xiao A, Pollmann SI, Grol MW, Beach RJ, Holdsworth DW, Sims SM, Darling MR, Dixon SJ. 2014. Loss of P2X7 nucleotide receptor function leads to abnormal fat distribution in mice. Purinergic Signal 10: 291–304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Perruzza L, Gargari G, Proietti M, Fosso B, D’Erchia AM, Faliti CE, Rezzonico-Jost T, Scribano D, Mauri L, Colombo D, Pellegrini G, Moregola A, Mooser C, Pesole G, Nicoletti M, Norata GD, Geuking MB, McCoy KD, Guglielmetti S, Grassi F. 2017. T Follicular Helper Cells Promote a Beneficial Gut Ecosystem for Host Metabolic Homeostasis by Sensing Microbiota-Derived Extracellular ATP. Cell Rep 18: 2566–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Donaldson GP, Lee SM, Mazmanian SK. 2016. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 14: 20–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gionchetti P, Rizzello F, Morselli C, Poggioli G, Tambasco R, Calabrese C, Brigidi P, Vitali B, Straforini G, Campieri M. 2007. High-dose probiotics for the treatment of active pouchitis. Dis Colon Rectum 50: 2075–82; discussion 82–4 [DOI] [PubMed] [Google Scholar]
  • 88.Hoermannsperger G, Clavel T, Hoffmann M, Reiff C, Kelly D, Loh G, Blaut M, Holzlwimmer G, Laschinger M, Haller D. 2009. Post-translational inhibition of IP-10 secretion in IEC by probiotic bacteria: impact on chronic inflammation. PLoS One 4: e4365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hormannsperger G, von Schillde MA, Haller D. 2013. Lactocepin as a protective microbial structure in the context of IBD. Gut Microbes 4: 152–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.von Schillde MA, Hormannsperger G, Weiher M, Alpert CA, Hahne H, Bauerl C, van Huynegem K, Steidler L, Hrncir T, Perez-Martinez G, Kuster B, Haller D. 2012. Lactocepin secreted by Lactobacillus exerts anti-inflammatory effects by selectively degrading proinflammatory chemokines. Cell Host Microbe 11: 387–96 [DOI] [PubMed] [Google Scholar]
  • 91.Castagliuolo I, Riegler MF, Valenick L, LaMont JT, Pothoulakis C. 1999. Saccharomyces boulardii protease inhibits the effects of Clostridium difficile toxins A and B in human colonic mucosa. Infect Immun 67: 302–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. 2013. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341: 569–73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Peng L, He Z, Chen W, Holzman IR, Lin J. 2007. Effects of butyrate on intestinal barrier function in a Caco-2 cell monolayer model of intestinal barrier. Pediatric research 61: 37–41 [DOI] [PubMed] [Google Scholar]
  • 94.Kim M, Qie Y, Park J, Kim CH. 2016. Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 20: 202–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lamas B, Richard ML, Leducq V, Pham HP, Michel ML, Da Costa G, Bridonneau C, Jegou S, Hoffmann TW, Natividad JM, Brot L, Taleb S, Couturier-Maillard A, Nion-Larmurier I, Merabtene F, Seksik P, Bourrier A, Cosnes J, Ryffel B, Beaugerie L, Launay JM, Langella P, Xavier RJ, Sokol H. 2016. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 22: 598–605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, Chao CC, Patel B, Yan R, Blain M, Alvarez JI, Kebir H, Anandasabapathy N, Izquierdo G, Jung S, Obholzer N, Pochet N, Clish CB, Prinz M, Prat A, Antel J, Quintana FJ. 2016. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med 22: 586–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.de Mello VD, Paananen J, Lindstrom J, Lankinen MA, Shi L, Kuusisto J, Pihlajamaki J, Auriola S, Lehtonen M, Rolandsson O, Bergdahl IA, Nordin E, Ilanne-Parikka P, Keinanen-Kiukaanniemi S, Landberg R, Eriksson JG, Tuomilehto J, Hanhineva K, Uusitupa M. 2017. Indolepropionic acid and novel lipid metabolites are associated with a lower risk of type 2 diabetes in the Finnish Diabetes Prevention Study. Sci Rep 7: 46337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, Krastel P, Schmitt EK, Omar AS, Creasey EA, Garner AL, Mohammadi S, O’Connell DJ, Abubucker S, Arthur TD, Franzosa EA, Huttenhower C, Murphy LO, Haiser HJ, Vlamakis H, Porter JA, Xavier RJ. 2017. Indoleacrylic Acid Produced by Commensal Peptostreptococcus Species Suppresses Inflammation. Cell Host Microbe 22: 25–37 e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jin UH, Lee SO, Sridharan G, Lee K, Davidson LA, Jayaraman A, Chapkin RS, Alaniz R, Safe S. 2014. Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Mol Pharmacol 85: 777–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, Diefenbach A. 2011. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334: 1561–5 [DOI] [PubMed] [Google Scholar]
  • 101.Lee JS, Cella M, McDonald KG, Garlanda C, Kennedy GD, Nukaya M, Mantovani A, Kopan R, Bradfield CA, Newberry RD, Colonna M. 2011. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat Immunol 13: 144–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, Wilhelm C, Veldhoen M. 2011. Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 147: 629–40 [DOI] [PubMed] [Google Scholar]
  • 103.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L. 2013. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39: 372–85 [DOI] [PubMed] [Google Scholar]
  • 104.Sokol H, Conway KL, Zhang M, Choi M, Morin B, Cao Z, Villablanca EJ, Li C, Wijmenga C, Yun SH, Shi HN, Xavier RJ. 2013. Card9 mediates intestinal epithelial cell restitution, T-helper 17 responses, and control of bacterial infection in mice. Gastroenterology 145: 591–601 e3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, Le A, Cowan TM, Nolan GP, Fischbach MA, Sonnenburg JL. 2017. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 551: 648–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo MR, Phillips RS, Fleet JC, Kortagere S, Mukherjee P, Fasano A, Le Ven J, Nicholson JK, Dumas ME, Khanna KM, Mani S. 2014. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 41: 296–310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Cuskin F, Lowe EC, Temple MJ, Zhu Y, Cameron EA, Pudlo NA, Porter NT, Urs K, Thompson AJ, Cartmell A, Rogowski A, Hamilton BS, Chen R, Tolbert TJ, Piens K, Bracke D, Vervecken W, Hakki Z, Speciale G, Munoz-Munoz JL, Day A, Pena MJ, McLean R, Suits MD, Boraston AB, Atherly T, Ziemer CJ, Williams SJ, Davies GJ, Abbott DW, Martens EC, Gilbert HJ. 2015. Human gut Bacteroidetes can utilize yeast mannan through a selfish mechanism. Nature 517: 165–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck TS, Nunez G, Martens EC. 2016. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 167: 1339–53 e21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fan P, Li L, Rezaei A, Eslamfam S, Che D, Ma X. 2015. Metabolites of Dietary Protein and Peptides by Intestinal Microbes and their Impacts on Gut. Curr Protein Pept Sci 16: 646–54 [DOI] [PubMed] [Google Scholar]
  • 110.Uranchimeg D, Kim JH, Kim JY, Lee WT, Park KA, Batbaatar G, Tundevrentsen S, Amgalanbaatar D, Lee JE. 2010. Recovered changes in the spleen by agmatine treatment after transient cerebral ischemia. Anat Cell Biol 43: 44–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Aricioglu F, Regunathan S. 2005. Agmatine attenuates stress-and lipopolysaccharide-induced fever in rats. Physiol Behav 85: 370–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zhu S, Ashok M, Li J, Li W, Yang H, Wang P, Tracey KJ, Sama AE, Wang H. 2009. Spermine protects mice against lethal sepsis partly by attenuating surrogate inflammatory markers. Mol Med 15: 275–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Levy M, Thaiss CA, Zeevi D, Dohnalova L, Zilberman-Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, Pevsner-Fischer M, Shapiro H, Christ A, Harmelin A, Halpern Z, Latz E, Flavell RA, Amit I, Segal E, Elinav E. 2015. Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell 163: 1428–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Frei R, Ferstl R, Konieczna P, Ziegler M, Simon T, Rugeles TM, Mailand S, Watanabe T, Lauener R, Akdis CA, O’Mahony L. 2013. Histamine receptor 2 modifies dendritic cell responses to microbial ligands. J Allergy Clin Immunol 132: 194–204 [DOI] [PubMed] [Google Scholar]
  • 115.Craciun S, Balskus EP. 2012. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A 109: 21307–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Dambrova M, Latkovskis G, Kuka J, Strele I, Konrade I, Grinberga S, Hartmane D, Pugovics O, Erglis A, Liepinsh E. 2016. Diabetes is Associated with Higher Trimethylamine N-oxide Plasma Levels. Exp Clin Endocrinol Diabetes 124: 251–6 [DOI] [PubMed] [Google Scholar]
  • 117.Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. 2013. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 19: 576–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li XS, Levison BS, Hazen SL. 2015. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res 116: 448–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Romano KA, Martinez-Del Campo A, Kasahara K, Chittim CL, Vivas EI, Amador-Noguez D, Balskus EP, Rey FE. 2017. Metabolic, Epigenetic, and Transgenerational Effects of Gut Bacterial Choline Consumption. Cell Host Microbe 22: 279–90 e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kubinak JL, Round JL. 2012. Toll-like receptors promote mutually beneficial commensal-host interactions. PLoS pathogens 8: e1002785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Chu H, Mazmanian SK. 2013. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat Immunol 14: 668–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wolf AJ, Reyes CN, Liang W, Becker C, Shimada K, Wheeler ML, Cho HC, Popescu NI, Coggeshall KM, Arditi M, Underhill DM. 2016. Hexokinase Is an Innate Immune Receptor for the Detection of Bacterial Peptidoglycan. Cell 166: 624–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Saha S, Jing X, Park SY, Wang S, Li X, Gupta D, Dziarski R. 2010. Peptidoglycan recognition proteins protect mice from experimental colitis by promoting normal gut flora and preventing induction of interferon-gamma. Cell Host Microbe 8: 147–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bank S, Skytt Andersen P, Burisch J, Pedersen N, Roug S, Galsgaard J, Ydegaard Turino S, Brodersen JB, Rashid S, Kaiser Rasmussen B, Avlund S, Bastholm Olesen T, Jurgen Hoffmann H, Kragh Thomsen M, Ostergaard Thomsen V, Frydenberg M, Andersen Nexo B, Sode J, Vogel U, Andersen V. 2014. Polymorphisms in the inflammatory pathway genes TLR2, TLR4, TLR9, LY96, NFKBIA, NFKB1, TNFA, TNFRSF1A, IL6R, IL10, IL23R, PTPN22, and PPARG are associated with susceptibility of inflammatory bowel disease in a Danish cohort. PLoS One 9: e98815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Guo C, Zhang L, Nie L, Zhang N, Xiao D, Ye X, Ou M, Liu Y, Zhang B, Wang M, Lin H, Yang G, Jing C. 2016. Association of polymorphisms in the MyD88, IRAK4 and TRAF6 genes and susceptibility to type 2 diabetes mellitus and diabetic nephropathy in a southern Han Chinese population. Mol Cell Endocrinol 429: 114–9 [DOI] [PubMed] [Google Scholar]
  • 126.Misch EA, Hawn TR. 2008. Toll-like receptor polymorphisms and susceptibility to human disease. Clinical science 114: 347–60 [DOI] [PubMed] [Google Scholar]
  • 127.Mowat AM. 2010. Does TLR2 regulate intestinal inflammation? Eur J Immunol 40: 318–20 [DOI] [PubMed] [Google Scholar]
  • 128.Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, Huot L, Grandjean T, Bressenot A, Delanoye-Crespin A, Gaillot O, Schreiber S, Lemoine Y, Ryffel B, Hot D, Nunez G, Chen G, Rosenstiel P, Chamaillard M. 2013. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest 123: 700–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Caricilli AM, Picardi PK, de Abreu LL, Ueno M, Prada PO, Ropelle ER, Hirabara SM, Castoldi A, Vieira P, Camara NO, Curi R, Carvalheira JB, Saad MJ. 2011. Gut Microbiota Is a Key Modulator of Insulin Resistance in TLR 2 Knockout Mice. PLoS biology 9: e1001212. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 130.Jiang W, Wang X, Zeng B, Liu L, Tardivel A, Wei H, Han J, MacDonald HR, Tschopp J, Tian Z, Zhou R. 2013. Recognition of gut microbiota by NOD2 is essential for the homeostasis of intestinal intraepithelial lymphocytes. J Exp Med 210: 2465–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Secher T, Normand S, Chamaillard M. 2013. NOD2 prevents emergence of disease-predisposing microbiota. Gut Microbes 4: 353–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, Sitaraman SV, Knight R, Ley RE, Gewirtz AT. 2010. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 328: 228–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Tang AT, Choi JP, Kotzin JJ, Yang Y, Hong CC, Hobson N, Girard R, Zeineddine HA, Lightle R, Moore T, Cao Y, Shenkar R, Chen M, Mericko P, Yang J, Li L, Tanes C, Kobuley D, Vosa U, Whitehead KJ, Li DY, Franke L, Hart B, Schwaninger M, Henao-Mejia J, Morrison L, Kim H, Awad IA, Zheng X, Kahn ML. 2017. Endothelial TLR4 and the microbiome drive cerebral cavernous malformations. Nature 545: 305–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, Paley MA, Antenus M, Williams KL, Erikson J, Wherry EJ, Artis D. 2012. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 37: 158–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. 2010. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature medicine 16: 228–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Olsen I, Jantzen E. 2001. Sphingolipids in Bacteria and Fungi. Anaerobe 7: 103–12 [Google Scholar]
  • 137.Matsuda JL, Gapin L, Fazilleau N, Warren K, Naidenko OV, Kronenberg M. 2001. Natural killer T cells reactive to a single glycolipid exhibit a highly diverse T cell receptor beta repertoire and small clone size. Proc Natl Acad Sci U S A 98: 12636–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.An D, Oh SF, Olszak T, Neves JF, Avci FY, Erturk-Hasdemir D, Lu X, Zeissig S, Blumberg RS, Kasper DL. 2014. Sphingolipids from a symbiotic microbe regulate homeostasis of host intestinal natural killer T cells. Cell 156: 123–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Mazmanian SK, Round JL, Kasper DL. 2008. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453: 620–5 [DOI] [PubMed] [Google Scholar]
  • 140.Ochoa-Reparaz J, Mielcarz DW, Wang Y, Begum-Haque S, Dasgupta S, Kasper DL, Kasper LH. 2010. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal immunology 3: 487–95 [DOI] [PubMed] [Google Scholar]
  • 141.Round JL, Mazmanian SK. 2010. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proceedings of the National Academy of Sciences of the United States of America 107: 12204–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Underhill D, Braun J. 2008. Current understanding of fungal microflora in inflammatory bowel disease pathogenesis. Inflammatory bowel diseases 14: 1147–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Franchimont D, Vermeire S, El Housni H, Pierik M, Van Steen K, Gustot T, Quertinmont E, Abramowicz M, Van Gossum A, Deviere J, Rutgeerts P. 2004. Deficient host-bacteria interactions in inflammatory bowel disease? The toll-like receptor (TLR)-4 Asp299gly polymorphism is associated with Crohn’s disease and ulcerative colitis. Gut 53: 987–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Wagener J, Malireddi RK, Lenardon MD, Koberle M, Vautier S, MacCallum DM, Biedermann T, Schaller M, Netea MG, Kanneganti TD, Brown GD, Brown AJ, Gow NA. 2014. Fungal chitin dampens inflammation through IL-10 induction mediated by NOD2 and TLR9 activation. PLoS Pathog 10: e1004050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Plato A, Hardison SE, Brown GD. 2015. Pattern recognition receptors in antifungal immunity. Semin Immunopathol 37: 97–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Jiang TT, Shao TY, Ang WXG, Kinder JM, Turner LH, Pham G, Whitt J, Alenghat T, Way SS. 2017. Commensal Fungi Recapitulate the Protective Benefits of Intestinal Bacteria. Cell Host Microbe 22: 809–16 e4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Vecchiarelli A, Pericolini E, Gabrielli E, Kenno S, Perito S, Cenci E, Monari C. 2013. Elucidating the immunological function of the Cryptococcus neoformans capsule. Future Microbiol 8: 1107–16 [DOI] [PubMed] [Google Scholar]
  • 148.Piccioni M, Monari C, Kenno S, Pericolini E, Gabrielli E, Pietrella D, Perito S, Bistoni F, Kozel TR, Vecchiarelli A. 2013. A purified capsular polysaccharide markedly inhibits inflammatory response during endotoxic shock. Infect Immun 81: 90–8 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES